WO2009102789A2 - Utilisation d'agonistes de rxr pour le traitement de l'arthrose - Google Patents

Utilisation d'agonistes de rxr pour le traitement de l'arthrose Download PDF

Info

Publication number
WO2009102789A2
WO2009102789A2 PCT/US2009/033795 US2009033795W WO2009102789A2 WO 2009102789 A2 WO2009102789 A2 WO 2009102789A2 US 2009033795 W US2009033795 W US 2009033795W WO 2009102789 A2 WO2009102789 A2 WO 2009102789A2
Authority
WO
WIPO (PCT)
Prior art keywords
rxr
cartilage
treatment
expression
osteoarthritis
Prior art date
Application number
PCT/US2009/033795
Other languages
English (en)
Other versions
WO2009102789A3 (fr
Inventor
Sunil Nagpal
Zhiyong Yang
Elisabeth Morris
Edward Lavallie
Lisa A. Collins-Racie
Original Assignee
Wyeth
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Wyeth filed Critical Wyeth
Publication of WO2009102789A2 publication Critical patent/WO2009102789A2/fr
Publication of WO2009102789A3 publication Critical patent/WO2009102789A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/202Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids having three or more double bonds, e.g. linolenic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/203Retinoic acids ; Salts thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis

Definitions

  • the present invention relates to methods of treating or preventing osteoarthritis with RXR agonists.
  • Osteoarthritis also known as degenerative joint disease, is characterized by degeneration of articular cartilage as well as proliferation and remodeling of subchondral bone. The usual symptoms are stiffness, limitation of motion, and pain. Osteoarthritis is the most common form of arthritis, and prevalence rates increase markedly with age.
  • osteoarthritis treatment approaches include exercise, medicines, rest and joint care, surgery, pain relief techniques, alternative therapies, and weight control.
  • the commonly used medicines in treating osteoarthritis include nonsteroidal anti-inflammatory drugs (NSAIDs), for example, aspirin, ibuprofen, naproxen sodium, ketoprofen; topical pain-relieving creams, rubs, and sprays (for example, capsaicin cream) applied directly to the skin; corticosteroids, typically injected into affected joints to relieve pain temporarily; and hyaluronic acid.
  • NSAIDs nonsteroidal anti-inflammatory drugs
  • corticosteroids typically injected into affected joints to relieve pain temporarily
  • hyaluronic acid for example, capsaicin cream
  • Surgery may be performed to resurface (smooth out) bones, reposition bones, and replace joints.
  • Retinoid X receptors are members of a large superfamily of intracellular hormone receptors. These proteins bind to specific DNA sequences and directly regulate transcription of target genes in response to activation by their specific ligands (Leid et al., Trends Biochem. Sci. 17:427-33 (1992); Leid et al., Cell 68:377-95 (1992); Mangelsdorf et al., Nature 345:224-29 (1990); and Yu et al., Cell 67:1251-66 (1991)).
  • the RXRs belong to a large subgroup of the superfamily defined by a conserved subregion within the DNA binding domain.
  • This group also includes the receptors for retinoic acid, thyroid hormone, and vitamin D as well as a number of other less well characterized proteins, called orphan receptors, that do not have known ligands.
  • the members of this class can bind to sequences related to the hexameric consensus AGGTCA.
  • RXR homodimers bind to tandem repeats of this consensus separated by a single base pair (Manglesdorf et al., Cell 66:555-61 (1991)), and apparently to additional elements including ⁇ -RARE (Zhang et al., Nature
  • RXR Mangelsdorf et al., Genes Dev. 6:329-44 (1992)).
  • Liver X receptors (LXRs), originally identified from liver as orphan receptors, are members of the nuclear hormone receptor super family and have been found to be negative regulators of macrophage inflammatory gene expression (see Published U.S. Patent Application No. 2004/0259948; Joseph SB et al., Nat. Med. 9:213-19 (2003)). LXRs are ligand-activated transcription factors and bind to DNA as obligate heterodimers with retinoid X receptors. While LXR ⁇ is restricted to certain tissues such as liver, kidney, adipose, intestine, and macrophages, LXR ⁇ displays a ubiquitous tissue distribution pattern.
  • LXRs Activation of LXRs by oxysterols (endogenous ligands) in macrophages results in the expression of several genes involved in lipid metabolism and reverse cholesterol transport, including ABCA1 , ABCG1 , and apolipoprotein E.
  • One aspect is for a method for the treatment of a mammal suffering from osteoarthritis comprising administering to the mammal in need thereof an RXR- responsive gene expression-modulating amount of an RXR agonist.
  • Another aspect is for a method for the treatment of a mammal suffering from osteoarthritis comprising administering to the mammal in need thereof an effective amount of an RXR agonist to relieve pain in osteoarthritic joints.
  • a further aspect is for a method of indentifying an RXR ligand capable of reducing an osteoarthritic effect in cartilage comprising: (a) providing a sample containing RXR; (b) contacting the sample with a test compound; and (c) determining whether the test compound reduces an osteoarthritic effect in cartilage.
  • Fig. 1 Expression of selected human nuclear receptors in articular cartilage from subjects with osteoarthritis compared to normal cartilage. mRNA levels for nuclear receptors judged to be expressed ("present") in HG-U95Av2 Affymetrix GeneChip® data of articular cartilage from severe OA patients. Values on the Y- axis reflect transcript levels measured on GeneChips® and expressed in parts per million (ppm).
  • LXR liver X receptor
  • RXR retinoid X receptor
  • RAR retinoic acid receptor
  • Rev Rev-erb
  • GR glucocorticoid receptor
  • EAR v-erbA-related
  • COU chicken ovalbumin upstream promoter transcription factor
  • CAR constitutive androstane receptor
  • PXR pregnane X receptor
  • MR mineralocorticoid receptor
  • SF steroidogenic factor
  • TR thyroid hormone receptor
  • NOR neuron-derived orphan receptor
  • Nurr Nur-related
  • SHP small heterodimer partner
  • FXR farnesoid X receptor.
  • Bars represent the mean of replicate qRT-PCR reactions ⁇ SEM * p ⁇ 0.05, ** p ⁇ 0.01 , comparison of all OA samples to normal samples; # p ⁇ 0.05, ## p ⁇ 0.01 , comparison of lesional cartilage samples to normals; tt p ⁇ 0.01 , comparison of non-lesional cartilage samples to normals, or non-lesional cartilage samples to matched lesional cartilage samples, as indicated by brackets in the figure.
  • Fig. 3 Comparison of RXR ⁇ and RXR ⁇ nuclear receptor expression in non- lesional and lesional human osteoarthhtic articular cartilage compared to normal cartilage.
  • RNA samples using the human NR-TLDA expressed as mean RQ (fold- change) ⁇ SEM for that cohort compared to normal sample Control 1 following normalization to the GUSB ( ⁇ -glucuronidase) endogenous control. ** p ⁇ 0.05 by Welch t test for both lesional OA vs. normal and non-lesional OA vs. normal comparisons.
  • Fig. 4 Primary OA chondrocytes down regulate RXR ⁇ and RXRy in response to treatment with IL-1 ⁇ or TNF ⁇ .
  • RNA prepared from the cells following culture was assayed by qRT-PCR to measure the effect of cytokine treatment on the expression of (A) RXR ⁇ and (B) RXR ⁇ . Bars represent the individual average fold change in expression values for the cytokine-treated cultures for each donor compared to untreated cultures from the same donor, ⁇ SD. * * p ⁇ 0.01 vs. control by Welch t test.
  • RXR ⁇ and RXR ⁇ are expressed in normal, non-lesional osteoarthritic, and lesional (severe) osteoarthritic cartilages.
  • the transcriptional level of RXR ⁇ and RXR ⁇ are significantly decreased in cartilage from osteoarthritis patients compared to normals.
  • RXRy is also expressed in articular cartilage, and the expression of RXR ⁇ and RXRy in articular chondrocytes is significantly reduced by inflammatory cytokines 11-1 ⁇ (RXR ⁇ ) and TNF ⁇ (RXRy).
  • RXR biology is further complicated by the fact that some heterodimeric receptor complexes (e.g. LXRs, FXR, and PPARs) can be independently activated by either the RXR's ligand, the RXR partner's ligand, or by both; alternatively, other RXR heterodimeric receptor complexes require the partner's ligand for activation (e.g.
  • the term “about” or “approximately” means within 20%, preferably within 10%, and more preferably within 5% of a given value or range.
  • an RXR- responsive gene expression-inducing amount refers to the amount of an effector molecule that, when administered to a mammal in need, is effective to at least partially ameliorate or to at least partially prevent conditions related to osteoarthritis.
  • expression includes the process by which DNA is transcribed into mRNA and translated into polypeptides or proteins.
  • RXR Retinoid X Receptor
  • RXR ⁇ is ubiquitously expressed, while RXR ⁇ expression is limited to liver, kidney, spleen, placenta, epidermis, and, as demonstrated herein, cartilage.
  • RXRy is expressed in muscle and brain, and, as demonstrated herein, cartilage.
  • GenBank® accession numbers for RXR ⁇ sequences include the following: human (Homo sapiens, NP_002948), mouse (Mus musculus, NP_035435, AAB36777, AAB36778), rat (Rattus norvegicus, NP 036937), orangutan (Pongo abelii, NP_001125717), zebrafish ⁇ Danio rerio, NP_571228, A2T929), frog (Xenopus laevis, P51128).
  • GenBank® accession numbers for RXR ⁇ sequences include the following: human (Homo sapiens, NP_068811), mouse (Mus musculus, NP_035436, BAA04859), rat (Rattus norvegicus, NP_996731), cow (Bos taurus, NP_001077109), frog (Xenopus laevis, NP_001080936, NP_001081830), zebrafish (Danio rerio, NP 571350, NP_571313, Q90415), dog (Canis lupus familiaris, Q5TJF7).
  • GenBank® accession numbers for RXRy sequences include the following: human (Homo sapiens, NP 008848, NP 001009598), mouse (Mus musculus, NP_033133), rat (Rattus norvegicus, NP_113953), cow (Bos taurus, N P_001068876), chicken (Gallus gallus, NP_990625), zebrafish (Danio rerio, NP_571292, Q6DHP9), orangutan (Pongo abelii, N P_001124824), pig (Sus scrofa, N P_001123685), frog (Xenopus laevis, P51129).
  • Liver X receptor refers to both LXR ⁇ and LXR ⁇ , and variants, isoforms, and active fragments thereof.
  • LXR ⁇ is ubiquitously expressed, while LXR ⁇ expression is limited to liver, kidney, intestine, spleen, adipose tissue, macrophages, skeletal muscle, and, as demonstrated herein, cartilage.
  • GenBank® accession numbers for LXR ⁇ sequences include the following: human (Homo sapiens, NP_005684, NP 001123573, NP 001123574), mouse (Mus musculus, NP_038867), rat (Rattus norvegicus, NP_113815), cow (Bos taurus, NP_001014861), pig (Sus scrofa, N P O01095284), chicken (Gallus gallus, NP_989873).
  • GenBank® accession numbers for LXR ⁇ include the following: human (Homo sapiens, NP 009052), mouse (M ⁇ s musculus, NP_033499), rat (Rattus norvegicus, Q62755), cow ⁇ Bos taurus, Q5BIS6).
  • the term "mammal” refers to a human, a non-human primate, canine, feline, bovine, ovine, porcine, murine, or other veterinary or laboratory mammal. Those skilled in the art recognize that a therapy which reduces the severity of a pathology in one species of mammal is predictive of the effect of the therapy on another species of mammal.
  • modulate encompasses either a decrease or an increase in activity or expression depending on the target molecule.
  • an RXR ⁇ modulator is considered to modulate the expression or activity of RXR ⁇ if the presence of such RXR ⁇ modulator results in an increase or decrease in RXR ⁇ expression or activity.
  • RXR agonists useful in the present invention include, but are not limited to, compounds that preferentially activate RXR over RAR (i.e. RXR specific agonists) and compounds that activate both RXR and RAR (i.e. pan agonists). It also includes compounds that activate RXR in a certain cellular context but not others (i.e. partial agonists). Representative compounds include those disclosed in U.S. Patent Nos. 5,399,586, 5,466,861 , 5,801 ,253, 6,506,917, 5,780,676, 5,962,731 , 6,320,074, 5,972,881, 5,770,378, and 5,721 ,103, and in Boehm et al., J. Med. Chem.
  • Pan agonists include, but are not limited to, 9-c/s retinoic acid, docosahexanoic acid, and phytanic acid.
  • Useful synthetic agonists include LG100268 (6-[1-(3,5,5,8,8-pentamethyl-5,6,7,8- tetrahydronaphthalen-2- yl)cyclopropyl]pyridine-3-carboxylic acid) and bexarotene (4-[1 -(5,6,7,8 - tetrahydro-3,5,5,8,8-pentamethyl-2-naphthalenyl) ethenyl] benzoic acid).
  • LXR agonists useful in the present invention include natural oxysterols, synthetic oxysterols, synthetic nonoxysterols, and natural nonoxysterols.
  • Exemplary natural oxysterols include 20(S) hydroxycholesterol, 22(R) hydroxycholesterol, 24(S) hydroxycholesterol, 25-hydroxycholesterol, 24(S), 25 epoxycholesterol, and 27-hydroxycholesterol.
  • Exemplary synthetic oxysterols include N,N-dimethyl-3 ⁇ -hydroxycholenamide (DMHCA).
  • Exemplary synthetic nonoxysterols include N-(2,2,2-trifluoroethyl)-N- ⁇ 4-[2,2,2-trifluoro-1 -hydroxy-1 - (trifluoromethyl)ethyl]phenyl ⁇ benzene sulfonamide (TO901317; Tularik 0901317), [3-(3-(2-chloro-trifluoromethylbenzyl-2,2- diphenylethylamino)propoxy)phenylacetic acid] (GW3965), N-methyl-N-[4-(2,2,2- trifluoro-1 -hydroxy-1 -trifluoromethyl-1 -ethyl)-phenyl]-benzenesulfonamide (TO314407), 4,5-dihydro-1-(3-(3-trifluoromethyl-7-propyl-benzisoxazol-6- yloxy)propyl)-2,6-pyrimidinedione, 3-chloro-4-(3-(7-
  • LXR agonists are disclosed, for example, in Published U.S. Patent Application Nos. 2006/0030612, 2005/0131014, 2005/0036992,
  • RXR activity is stimulated in a cell by contacting the cell with an RXR agonist.
  • RXR agonists are described above in Section II.
  • Other RXR agonists that can be used to stimulate the RXR activity can be identified using screening assays that select for such compounds, as described in detail herein (Section V).
  • Modulatory methods can be performed in vitro (e.g., by culturing the cell with an RXR agonist or by introducing an RXR agonist into cells in culture) or, alternatively, in vivo (e.g., by administering an RXR agonist to a subject or by introducing an RXR agonist into cells of a subject).
  • cells can be obtained from a subject by standard methods and incubated (i.e., cultured) in vitro with an RXR agonist to modulate RXR activity in the cells.
  • the invention provides a method for preventing osteoarthritis in a subject by administering to the subject an RXR agonist.
  • Administration of a prophylactic RXR agonist can occur prior to the manifestation of osteoarthritis symptoms, such that osteoarthritis is prevented or, alternatively, delayed in its progression.
  • a modulatory method of the invention involves contacting a cell with an RXR agonist.
  • modulatory methods can be performed in vitro (e.g., by culturing the cell with an RXR agonist) or, alternatively, in vivo (e.g., by administering an RXR agonist to a subject).
  • RXR agonists can also be useful for treating pain in osteoarthritic joints.
  • RXR agonists can be effective in treating acute pain (short duration) or chronic pain (regularly reoccurring or persistent) associated with osteoarthritis.
  • RXR agonists are administered to subjects in a biologically compatible form suitable for pharmaceutical administration in vivo.
  • biologically compatible form suitable for administration in vivo is meant a form of the RXR agonist to be administered in which any toxic effects are outweighed by the therapeutic effects of the agonist.
  • subject is intended to include living organisms in which an immune response can be elicited, for example, mammals.
  • Administration of RXR agonists as described herein can be in any pharmacological form including a therapeutically effective amount of an RXR agonist alone or in combination with a pharmaceutically acceptable carrier.
  • a therapeutically effective amount of an RXR agonist may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the RXR agonist to elicit a desired response in the individual. Dosage regime may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily, or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation.
  • the therapeutic or pharmaceutical compositions of the present invention can be administered by any suitable route known in the art including, for example, oral, intravenous, subcutaneous, intramuscular, transdermal, intrathecal, or intracerebral or administration to cells in ex vivo treatment protocols. Administration can be either rapid as by injection or over a period of time as by slow infusion or administration of slow release formulation. For treating or preventing osteoarthritis, administration of the therapeutic or pharmaceutical compositions of the present invention can be performed, for example, by oral administration or by intra-articular injection.
  • RXR agonists can be stably linked to a polymer such as polyethylene glycol to obtain desirable properties of solubility, stability, half-life, and other pharmaceutically advantageous properties (see, e.g., Davis et ai, Enzyme Eng. 4:169-73 (1978); Burnham NL, Am. J. Hosp. Pharm. 51 :210-18 (1994)).
  • RXR agonists can be in a composition that aids in delivery into the cytosol of a cell.
  • an RXR agonist may be conjugated with a carrier moiety such as a liposome that is capable of delivering the agonist into the cytosol of a cell.
  • a carrier moiety such as a liposome that is capable of delivering the agonist into the cytosol of a cell.
  • an RXR agonist can be delivered directly into a cell by microinjection.
  • RXR agonists can be employed in the form of pharmaceutical preparations. Such preparations are made in a manner well known in the pharmaceutical art. One preferred preparation utilizes a vehicle of physiological saline solution, but it is contemplated that other pharmaceutically acceptable carriers such as physiological concentrations of other non-toxic salts, five percent aqueous glucose solution, sterile water or the like may also be used.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. The use of such media and agents for pharmaceutically active substances is well known in the art.
  • any conventional media or agent is incompatible with the RXR agonist
  • use thereof in the therapeutic compositions is contemplated.
  • Supplementary active compounds can also be incorporated into the compositions. It may also be desirable that a suitable buffer be present in the composition.
  • Such solutions can, if desired, be lyophilized and stored in a sterile ampoule ready for reconstitution by the addition of sterile water for ready injection.
  • the primary solvent can be aqueous or alternatively non-aqueous.
  • RXR agonists can also be incorporated into a solid or semi-solid biologically compatible matrix which can be implanted into tissues requiring treatment.
  • the carrier can also contain other pharmaceutically-acceptable excipients for modifying or maintaining the pH, osmolarity, viscosity, clarity, color, sterility, stability, rate of dissolution, or odor of the formulation.
  • Dose administration can be repeated depending upon the pharmacokinetic parameters of the dosage formulation and the route of administration used.
  • formulations containing RXR agonists are to be administered orally.
  • Such formulations are preferably encapsulated and formulated with suitable carriers in solid dosage forms.
  • suitable carriers, excipients, and diluents include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, gelatin, syrup, methyl cellulose, methyl- and propylhydroxybenzoates, talc, magnesium, stearate, water, mineral oil, and the like.
  • the formulations can additionally include lubricating agents, wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents, or flavoring agents.
  • the compositions may be formulated so as to provide rapid, sustained, or delayed release of the active ingredients after administration to the patient by employing procedures well known in the art.
  • the formulations can also contain substances that diminish proteolytic degradation and/or substances which promote absorption such as, for example, surface active agents.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the RXR agonist and the particular therapeutic effect to be achieved and (b) the limitations inherent in the art of compounding such an active compound for the treatment of OA in individuals.
  • the specific dose can be readily calculated by one of ordinary skill in the art, e.g., according to the approximate body weight or body surface area of the patient or the volume of body space to be occupied. The dose will also be calculated dependent upon the particular route of administration selected. Further refinement of the calculations necessary to determine the appropriate dosage for treatment is routinely made by those of ordinary skill in the art. Such calculations can be made without undue experimentation by one skilled in the art in light of the RXR agonist activities disclosed herein in assay preparations of target cells. Exact dosages are determined in conjunction with standard dose-response studies.
  • the amount of the composition actually administered will be determined by a practitioner, in the light of the relevant circumstances including the condition or conditions to be treated; the choice of composition to be administered; the age, weight, and response of the individual patient; the severity of the patient's symptoms; and the chosen route of administration.
  • RXR agonists Toxicity and therapeutic efficacy of such RXR agonists can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, for example, for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 50 ZED 50 .
  • RXR agonists that exhibit large therapeutic indices are preferred. While RXR agonists that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such agonists to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such RXR agonists lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (i.e., the concentration of RXR agonist that achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • IC 50 i.e., the concentration of RXR agonist that achieves a half-maximal inhibition of symptoms
  • levels in plasma may be measured, for example, by high performance liquid chromatography.
  • articular chondrocytes can be isolated and RNA prepared and analyzed for the levels of expression of TN Fa and other genes implicated in osteoarthritis.
  • the levels of gene expression i.e., a gene expression pattern
  • the gene expression pattern can serve as a marker, indicative of the physiological response of the cells to the RXR agonist. Accordingly, this response state may be determined before, and at various points during, treatment of the individual with the RXR agonist.
  • compositions containing RXR agonists can be administered exogenously, and it would likely be desirable to achieve certain target levels of RXR agonist in sera, in any desired tissue compartment, and/or in the affected tissue. It would, therefore, be advantageous to be able to monitor the levels of RXR agonist in a patient or in a biological sample including a tissue biopsy sample obtained from a patient. Accordingly, the present invention also provides methods for detecting the presence of RXR agonist in a sample from a patient. V. Screening Assays
  • expression levels of RXR-responsive genes or activity levels of proteins therefrom can be used to facilitate design and/or identification of compounds that treat osteoarthritis through an RXR-based mechanism. Accordingly, the invention provides methods (also referred to herein as
  • screening assays for identifying RXR agonists.
  • Compounds thus identified can be used in the treatment of osteoarthritis as described elsewhere herein.
  • Test compounds can be obtained, for example, using any of the numerous approaches in combinatorial library methods known in the art, including spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the 'one-bead one-compound' library method; and synthetic library methods using affinity chromatography selection.
  • An exemplary screening assay is a cell-based assay in which a cell that expresses RXR is contacted with a test compound, and the ability of the test compound to treat an osteoarthritic condition through an RXR-based mechanism. Determining the ability of the test compound to treat an osteoarthritic condition can be accomplished by monitoring, for example, DNA, mRNA, or protein levels, or by measuring the levels of activity of, e.g., TNF ⁇ , all by methods well known to those of ordinary skill in the art.
  • the cell for example, can be of mammalian origin, e.g., human.
  • Novel modulators identified by the above-described screening assays can be used for treatments as described herein.
  • IL-1 ⁇ and TNF ⁇ were purchased from R&D Systems (Minneapolis, MN).
  • RNA Isolation of RNA from primary cartilage tissue and from chondrocytes in culture
  • the OA cartilage samples were obtained as whole joints within 2 hours of surgery, and the articular cartilage was shaved from the joint surfaces taking great care to avoid any pannus, fibrotic tissues, subchondral bone, and other non-cartilaginous regions of the joint.
  • Non-osteoarthritic cartilage samples were obtained from individuals without a clinical diagnosis or symptoms of OA, and the specimens were evaluated histologically to confirm the classification prior to inclusion in this study.
  • Cartilage pieces were flash-frozen in liquid nitrogen and stored at -80 0 C until processed for RNA isolation.
  • the frozen cartilage was pulverized using a Spex Certiprep freezer mill Model 6750 at 15 Hz twice for 1 minute each under liquid nitrogen.
  • the frozen powdered cartilage was resuspended in ice-cold 4M guanidinium isothiocyanate (GITC) (Invitrogen,
  • RNA was subsequently precipitated by the addition of isopropanol, and further purified using an RNeasy Mini Kit (Qiagen, Valencia, CA) according to the manufacturer's protocol. RNA quantity and purity was measured by ultraviolet absorbance at A260/A280, and RNA quality was assessed by the RNA6000 assay using the Agilent BioAnalyzer 2100 (Palo Alto, CA). RNA yields averaged between 5-10 mg of total RNA per gram of cartilage tissue.
  • RNA was subsequently prepared using TRIzol reagent (Invitrogen) according to the manufacturer's protocol. Primary chondrocytes in monolayer culture were lysed by direct addition of TRIzol reagent followed by standard TRIzol RNA purification methodologies. Chondrocyte cell culture
  • Chondrocytes were isolated from fresh human articular cartilage using a standard method previously described (Heinlein et al., Endocr. Rev. 25:276-308 (2004)). Cells were cultured in 10% FBS containing DMEM/F12 growth media for 2-3 days in 12 well culture plates at a density of 1-2 x 10 6 cells/well. Chondrocyte cultures were stimulated with cytokines (TNF ⁇ : 10 ng/ml; IL-1 ⁇ : 1 ng/ml) for 18 hours.
  • cytokines TNF ⁇ : 10 ng/ml
  • IL-1 ⁇ 1 ng/ml
  • the fragmented cRNAs were diluted in 1x MES buffer containing 100 ⁇ g/ml herring sperm DNA and 500 ⁇ g/ml acetylated BSA and denatured for 5 min at 99°C followed immediately by 5 min at 45°C. Insoluble material was removed from the hybridization mixture by a brief centrifugation, and the hybridization mix was added to each array and incubated at 45°C for 16 hr with continuous rotation at 60 rpm. After incubation, the hybridization mix was removed and the chips were extensively washed and stained with Streptavidin R-phycoerythrin (Molecular Probes, Eugene, OR) using the GeneChip® Fluidics Station 400 following the manufacturer's specifications. The raw florescent intensity value of each transcript was measured at a resolution of 6 microns with a Hewlett-Packard Gene Array Scanner.
  • cDNA synthesis and Quantitative RT-PCR (TagMan®) cDNA was prepared from purified RNA using the High-Capacity cDNA
  • RNA for TaqMan® analysis was purified from dissected and frozen cartilage tissue as described above, followed by two more rounds of phenol/chloroform extraction followed by RNeasy (Qiagen) column binding and elution.
  • Pre-designed TaqMan® probe/primer assay sets were obtained for the following nuclear receptor genes: NR1 H3 (LXR ⁇ ), Hs00172885_m1 ; NR1 H2 (LXR ⁇ ), Hs00173195_m1 ; NR2B1 (RXR ⁇ ), HS01067640_m1 ; and NR1 B3 (RARy), Hs00171273_m1.
  • GeneChip® software 3.2 (Affymetrix), which uses an algorithm to determine whether a gene is "present” or “absent”, as well as the specific hybridization intensity values or "average differences” of each gene on the array, was used to evaluate the gene chip data for all 49 identified human nuclear receptors (Robinson-Rechavi et al., Trends Genet. 17:554-56 (2001)). The average difference for each gene was normalized to frequency values by referral to the average differences of 11 control transcripts of known abundance that were spiked into each hybridization mix according to the procedure of Hill et al. (Science 290:809-12 (2000)).
  • Example 2 Quantitative RT-PCR experiments (qRT-PCR) were performed on cartilage RNA from a subset of the donors that were profiled by gene chip, chosen to represent cartilage with grossly severe lesions (83S and 86S), non- lesional cartilage from the same joints (83M and 86M), and cartilage from normal human joints (Control 1 and 2).
  • Applicants measured LXR ⁇ , LXR ⁇ , and RXR ⁇ by qRT-PCR to confirm the gene chip expression results and to investigate whether these members of LXR transcriptional complexes might be dysregulated in OA cartilage compared to normal.
  • the results, shown in Figures 2A-C confirmed that all three genes were expressed in articular cartilage.
  • Example 3 The differences in expression of RXR ⁇ and RXR ⁇ in non-lesional and lesional OA cartilage compared to normal, expressed as fold-change, are shown in Figure 3. Both RXR ⁇ (NR2B1) and RXR ⁇ (NR2B2) were found to be expressed at significantly lower levels in both non-lesional and lesional OA cartilage compared to normal.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

L'invention porte sur des procédés de prévention et de traitement l'arthrose par l'utilisation d'agonistes de RXR.
PCT/US2009/033795 2008-02-15 2009-02-11 Utilisation d'agonistes de rxr pour le traitement de l'arthrose WO2009102789A2 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US6595308P 2008-02-15 2008-02-15
US61/065,953 2008-02-15
US6991908P 2008-03-19 2008-03-19
US61/069,919 2008-03-19

Publications (2)

Publication Number Publication Date
WO2009102789A2 true WO2009102789A2 (fr) 2009-08-20
WO2009102789A3 WO2009102789A3 (fr) 2009-10-29

Family

ID=40626610

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/033795 WO2009102789A2 (fr) 2008-02-15 2009-02-11 Utilisation d'agonistes de rxr pour le traitement de l'arthrose

Country Status (2)

Country Link
US (1) US20090209601A1 (fr)
WO (1) WO2009102789A2 (fr)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011130359A1 (fr) * 2010-04-15 2011-10-20 Trustees Of Dartmouth College Films de nitrure de silicium améliorés et procédés
CA2858882C (fr) 2011-12-13 2021-02-02 Trustees Of Dartmouth College Traitement de trouble auto-immun a l'aide d'agonistes rxr
KR102605349B1 (ko) 2016-03-10 2023-11-22 아이오 테라퓨틱스, 인크. Rxr 작용제 및 갑상선 호르몬의 조합을 사용한 자가면역 질환의 치료
CA3076373A1 (fr) 2017-09-20 2019-03-28 Io Therapeutics, Inc. Traitement de maladie avec des esters d'agonistes de rxr selectifs
WO2022136344A1 (fr) * 2020-12-21 2022-06-30 Université Libre de Bruxelles Modulation de prdm12 à utiliser dans le traitement d'états douloureux
US11896558B2 (en) 2021-12-07 2024-02-13 Io Therapeutics, Inc. Use of an RXR agonist and taxanes in treating Her2+ cancers

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6136795A (en) * 1998-11-18 2000-10-24 Omni Nutraceuticals, Inc Dietary regimen of nutritional supplements for relief of symptoms of arthritis
WO2001080894A2 (fr) * 2000-04-20 2001-11-01 Allergan, Inc. Utilisation d'antagonistes ou d'agonistes du recepteur retinoide dans le traitement de pathologies des cartilages et os
US20040082655A1 (en) * 2002-10-23 2004-04-29 Parks L. Dean Method of treating musculoskeletal and connective tissue inflammations
US20060024356A1 (en) * 2004-07-01 2006-02-02 Nestec S.A. Canine osteoarthritis diet formulation
WO2007048510A1 (fr) * 2005-10-25 2007-05-03 Werner Bollag Agonistes et antagonistes rxr, seuls ou en conjonction avec des ligands ppar, dans le traitement des maladies métaboliques et cardiovasculaires
US20070141138A1 (en) * 2005-12-20 2007-06-21 Cenestra Llc Omega 3 fatty acid formulations
WO2008036239A2 (fr) * 2006-09-19 2008-03-27 Wyeth Utilisation d'agonistes lxr pour le traitement de l'ostéoarthrite

Family Cites Families (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US6992108B1 (en) * 1991-12-18 2006-01-31 The Salk Institute For Biological Studies Means for the modulation of processes mediated by retinoid receptors and compounds useful therefor
US6320074B1 (en) * 1992-04-22 2001-11-20 Ligand Pharmaceuticals Incorporated Compounds having selective activity for retinoid X receptors, and means for modulation of processes mediated by retinoid X receptors
US5962731A (en) * 1992-04-22 1999-10-05 Ligand Pharmaceuticals Incorporated Compounds having selective activity for retinoid X receptors, and means for modulation of processes mediated by retinoid X receptors
US5780676A (en) * 1992-04-22 1998-07-14 Ligand Pharmaceuticals Incorporated Compounds having selective activity for Retinoid X Receptors, and means for modulation of processes mediated by Retinoid X Receptors
US5466861A (en) * 1992-11-25 1995-11-14 Sri International Bridged bicyclic aromatic compounds and their use in modulating gene expression of retinoid receptors
US5399586A (en) * 1993-03-11 1995-03-21 Allergan, Inc. Treatment of mammals afflicted with tumors with compounds having RXR retinoid receptor agonist activity
WO1996005165A1 (fr) * 1994-08-10 1996-02-22 F. Hoffmann-La Roche Ag Ligands des recepteurs x de l'acide retinoique
US5721103A (en) * 1994-12-30 1998-02-24 Ligand Pharmaceuticals Incorporated Trienoic retinoid compounds and methods
US5770378A (en) * 1994-12-30 1998-06-23 Ligand Pharmaceuticals, Inc. Tricyclic retinoids, methods for their production and use
AU7074496A (en) * 1995-09-18 1997-04-09 Ligand Pharmaceuticals Incorporated Ppar gamma antagonists for treating obesity
US6316503B1 (en) * 1999-03-15 2001-11-13 Tularik Inc. LXR modulators
US20030086923A1 (en) * 1999-12-13 2003-05-08 Sparrow Carl P. Method for the prevention and/or treatment of atherosclerosis
JP5082033B2 (ja) * 2001-12-21 2012-11-28 エグゼリクシス パテント カンパニー エルエルシー Lxrのモジュレーター
MXPA04006203A (es) * 2001-12-21 2004-10-15 Pharmacia Corp Moduladores aromaticos del receptor x de higado de tioeter.
US7482366B2 (en) * 2001-12-21 2009-01-27 X-Ceptor Therapeutics, Inc. Modulators of LXR
US20050123580A1 (en) * 2002-02-28 2005-06-09 Burris Thomas P. Method of treating atherosclerosis and hypercholesterolemia
EP1511723A1 (fr) * 2002-05-24 2005-03-09 Pharmacia Corporation Modulateurs de recepteur x hepatique a base de sulfone
CA2486644A1 (fr) * 2002-05-24 2003-12-04 Pharmacia Corporation Modulateurs des recepteurs x du foie a structure anilino
JP2005539081A (ja) * 2002-09-17 2005-12-22 ファルマシア コーポレイション 芳香族肝臓x受容体モジュレーター
AU2003301216A1 (en) * 2002-12-23 2004-07-22 Irm Llc Novel use of liver x receptor agonists
US20040259948A1 (en) * 2003-01-10 2004-12-23 Peter Tontonoz Reciprocal regulation of inflammation and lipid metabolism by liver X receptors
US20050009837A1 (en) * 2003-05-20 2005-01-13 City Of Hope Modulators of lipid metabolism and methods of use
AU2004298486A1 (en) * 2003-12-12 2005-06-30 Wyeth Quinolines useful in treating cardiovascular disease
AR050279A1 (es) * 2004-08-03 2006-10-11 Wyeth Corp Indazoles como moduladores de lxr utiles en la preparacion de medicamentos pata el tratamiento de enfermedades cardiovasculares y mediadas por th1 y composiciones farmaceuticas que los contienen como principio activo.

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6136795A (en) * 1998-11-18 2000-10-24 Omni Nutraceuticals, Inc Dietary regimen of nutritional supplements for relief of symptoms of arthritis
WO2001080894A2 (fr) * 2000-04-20 2001-11-01 Allergan, Inc. Utilisation d'antagonistes ou d'agonistes du recepteur retinoide dans le traitement de pathologies des cartilages et os
US20040082655A1 (en) * 2002-10-23 2004-04-29 Parks L. Dean Method of treating musculoskeletal and connective tissue inflammations
US20060024356A1 (en) * 2004-07-01 2006-02-02 Nestec S.A. Canine osteoarthritis diet formulation
WO2007048510A1 (fr) * 2005-10-25 2007-05-03 Werner Bollag Agonistes et antagonistes rxr, seuls ou en conjonction avec des ligands ppar, dans le traitement des maladies métaboliques et cardiovasculaires
US20070141138A1 (en) * 2005-12-20 2007-06-21 Cenestra Llc Omega 3 fatty acid formulations
WO2008036239A2 (fr) * 2006-09-19 2008-03-27 Wyeth Utilisation d'agonistes lxr pour le traitement de l'ostéoarthrite

Also Published As

Publication number Publication date
US20090209601A1 (en) 2009-08-20
WO2009102789A3 (fr) 2009-10-29

Similar Documents

Publication Publication Date Title
Senf et al. FOXO signaling is required for disuse muscle atrophy and is directly regulated by Hsp70
Felipe et al. Modulation of resistin expression by retinoic acid and vitamin A status
Lin et al. Sirt2 suppresses inflammatory responses in collagen-induced arthritis
Mödder et al. Regulation of circulating sclerostin levels by sex steroids in women and in men
Villanueva et al. TLE3 is a dual-function transcriptional coregulator of adipogenesis
Caverzasio et al. Essential role of Wnt3a-mediated activation of mitogen-activated protein kinase p38 for the stimulation of alkaline phosphatase activity and matrix mineralization in C3H10T1/2 mesenchymal cells
Xing et al. Genetic evidence that thyroid hormone is indispensable for prepubertal insulin‐like growth factor–I expression and bone acquisition in mice
Lai et al. Cooperation of genomic and rapid nongenomic actions of estrogens in synaptic plasticity
US20090209601A1 (en) Use of rxr agonists for the treatment of osteoarthritis
US20050136429A1 (en) SIRT1 modulation of adipogenesis and adipose function
US20080070883A1 (en) Use of LXR modulators for the prevention and treatment of skin aging
Moult et al. Hormonal regulation of hippocampal dendritic morphology and synaptic plasticity
US20090012053A1 (en) Use of LXR agonists for the treatment of osteoarthritis
Collins-Racie et al. Global analysis of nuclear receptor expression and dysregulation in human osteoarthritic articular cartilage: reduced LXR signaling contributes to catabolic metabolism typical of osteoarthritis
Liu et al. WNT16 is upregulated early in mouse TMJ osteoarthritis and protects fibrochondrocytes against IL-1β induced inflammatory response by regulation of RUNX2/MMP13 cascade
Han et al. Fibromodulin is involved in autophagy and apoptosis of granulosa cells affecting the follicular atresia in chicken
Chen et al. Metformin impairs systemic bile acid homeostasis through regulating SIRT1 protein levels
Engeland et al. Sex Differences in Adrenal Bmal1 Deletion–Induced Augmentation of Glucocorticoid Responses to Stress and ACTH in Mice
Murahashi et al. Oral administration of EP4-selective agonist KAG-308 suppresses mouse knee osteoarthritis development through reduction of chondrocyte hypertrophy and TNF secretion
Yi et al. Tyrosyl phosphorylation of PZR promotes hypertrophic cardiomyopathy in PTPN11-associated Noonan syndrome with multiple lentigines
Gao et al. Interactions between nuclear receptors glucocorticoid receptor α and peroxisome proliferator–activated receptor α form a negative feedback loop
US11122997B2 (en) Modulating the aryl hydrocarbon receptor system to treat major depressive disorder
He et al. GPRC5B protects osteoarthritis by regulation of autophagy signaling
Diehl et al. Research resource: comparative nuclear receptor atlas: basal and activated peritoneal B-1 and B-2 cells
Xu et al. Sirt1 alleviates osteoarthritis via promoting FoxO1 nucleo-cytoplasm shuttling to facilitate autophagy

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09710117

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09710117

Country of ref document: EP

Kind code of ref document: A2